Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 34
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Cell Death Dis ; 6: e2011, 2015 Dec 03.
Artigo em Inglês | MEDLINE | ID: mdl-26633717

RESUMO

Serum response factor (SRF) is a transcription factor known to mediate phenotypic plasticity in smooth muscle cells (SMCs). Despite the critical role of this protein in mediating intestinal injury response, little is known about the mechanism through which SRF alters SMC behavior. Here, we provide compelling evidence for the involvement of SRF-dependent microRNAs (miRNAs) in the regulation of SMC apoptosis. We generated SMC-restricted Srf inducible knockout (KO) mice and observed both severe degeneration of SMCs and a significant decrease in the expression of apoptosis-associated miRNAs. The absence of these miRNAs was associated with overexpression of apoptotic proteins, and we observed a high level of SMC death and myopathy in the intestinal muscle layers. These data provide a compelling new model that implicates SMC degeneration via anti-apoptotic miRNA deficiency caused by lack of SRF in gastrointestinal motility disorders.


Assuntos
Mucosa Intestinal/metabolismo , MicroRNAs/metabolismo , Fator de Resposta Sérica/metabolismo , Animais , Apoptose , Diferenciação Celular , Proliferação de Células , Humanos , Intestinos/citologia , Intestinos/patologia , Camundongos , Miócitos de Músculo Liso , Transdução de Sinais
2.
J Biol Chem ; 276(36): 34175-81, 2001 Sep 07.
Artigo em Inglês | MEDLINE | ID: mdl-11447226

RESUMO

Retinoids block smooth muscle cell (SMC) proliferation and attenuate neointimal formation after vascular injury, presumably through retinoid receptor-mediated changes in gene expression. To identify target genes in SMC whose encoded proteins could contribute to such favorable biological effects, we performed a subtractive screen for retinoid-inducible genes in cultured SMC. Here, we report on the cloning and initial characterization of a novel retinoid-inducible serine carboxypeptidase (RISC). Expression of RISC is low in cultured SMC but progressively increases over a 5-day time-course treatment with all-trans-retinoic acid. A near full-length rat RISC cDNA was cloned and found to have a 452-amino acid open reading frame containing an amino-terminal signal sequence, followed by several conserved domains comprising the catalytic triad common to members of the serine carboxypeptidase family. In vitro transcription and translation experiments showed that the rat RISC cDNA generates an approximately 51-kDa protein. Confocal immunofluorescence microscopy of COS-7 cells transiently transfected with a RISC-His tag plasmid revealed cytosolic localization of the fusion protein. Western blotting studies using conditioned medium from transfected COS-7 cells suggest that RISC is a secreted protein. Tissue Northern blotting studies demonstrated robust expression of RISC in rat aorta, bladder, and kidney with much lower levels in all other tissues analyzed; high level RISC expression was also observed in human kidney. In situ hybridization verified the localization of RISC to medial SMC of the adult rat aorta. Interestingly, expression in kidney was restricted to proximal convoluted tubules; little or no expression was observed in glomerular cells, distal convoluted and collecting tubules, or medullary cells. Radiation hybrid mapping studies placed the rat RISC locus on chromosome 10q. These studies reveal a novel retinoid-inducible protease whose activity may be involved in vascular wall and kidney homeostasis.


Assuntos
Carboxiliases/genética , Carboxipeptidases/química , Carboxipeptidases/genética , Músculo Liso Vascular/metabolismo , Sequência de Aminoácidos , Animais , Aorta/metabolismo , Sequência de Bases , Northern Blotting , Western Blotting , Células COS , Carboxiliases/química , Carboxipeptidases/biossíntese , Células Cultivadas , Mapeamento Cromossômico , Clonagem Molecular , DNA Complementar/metabolismo , Hibridização In Situ , Rim/metabolismo , Masculino , Microscopia Confocal , Microscopia de Fluorescência , Dados de Sequência Molecular , Hibridização de Ácido Nucleico , Fases de Leitura Aberta , Fenótipo , Plasmídeos/metabolismo , Biossíntese de Proteínas , RNA Mensageiro/metabolismo , Mapeamento de Híbridos Radioativos , Ratos , Ratos Sprague-Dawley , Proteínas Recombinantes de Fusão/metabolismo , Homologia de Sequência de Aminoácidos , Fatores de Tempo , Distribuição Tecidual , Transcrição Gênica , Transfecção
3.
Biochem J ; 357(Pt 2): 587-92, 2001 Jul 15.
Artigo em Inglês | MEDLINE | ID: mdl-11439113

RESUMO

A hallmark of cultured smooth muscle cells (SMCs) is the rapid down-regulation of several lineage-restricted genes that define their in vivo differentiated phenotype. Identifying factors that maintain an SMC differentiated phenotype has important implications in understanding the molecular underpinnings governing SMC differentiation and their subversion to an altered phenotype in various disease settings. Here, we show that several G-protein coupled receptors [alpha-thrombin, lysophosphatidic acid and angiotensin II (AII)] increase the expression of smooth muscle calponin (SM-Calp) in rat and human SMC. The increase in SM-Calp protein appears to be selective for G-protein-coupled receptors as epidermal growth factor was without effect. Studies using AII showed a 30-fold increase in SM-Calp protein, which was dose- and time-dependent and mediated by the angiotensin receptor-1 (AT1 receptor). The increase in SM-Calp protein with AII was attributable to transcriptional activation of SM-Calp based on increases in steady-state SM-Calp mRNA, increases in SM-Calp promoter activity and complete abrogation of protein induction with actinomycin D. To examine the potential role of extracellular signal-regulated kinase (Erk1/2), protein kinase B, p38 mitogen-activated protein kinase and protein kinase C in AII-induced SM-Calp, inhibitors to each of the signalling pathways were used. None of these signalling molecules appears to be crucial for AII-induced SM-Calp expression, although Erk1/2 may be partially involved. These results identify SM-Calp as a target of AII-mediated signalling, and suggest that the SMC response to AII may incorporate a novel activity of SM-Calp.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação ao GTP/metabolismo , Regulação da Expressão Gênica/fisiologia , Músculo Liso Vascular/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Transcrição Gênica/fisiologia , Animais , Aorta , Diferenciação Celular , Células Cultivadas , Inibidores Enzimáticos/farmacologia , Flavonoides/farmacologia , Regulação da Expressão Gênica/efeitos dos fármacos , Genes Reporter , Humanos , Luciferases/genética , Proteínas dos Microfilamentos , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Proteínas Musculares/genética , Músculo Liso Vascular/citologia , Fosforilação , RNA Mensageiro/genética , Ratos , Ratos Endogâmicos WKY , Transcrição Gênica/efeitos dos fármacos , Transfecção , Proteínas Quinases p38 Ativadas por Mitógeno , Calponinas
5.
Mamm Genome ; 12(3): 187-91, 2001 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-11252166

RESUMO

Smooth muscle calponin (Cnn1) is a multifunctional protein whose expression is tightly restricted to differentiated smooth muscle cell (SMC) lineages during embryonic and post-natal life. As such, Cnn1 represents an ideal locus from which to dissect out regulatory elements that control its expression and hence the mature SMC phenotype. Previous work has focused on the expression and chromosomal mapping of the rat and human Cnn1 orthologs. In this report, we describe a unique pattern of Cnn1 expression during the growth and differentiation of BC3H1 cells, a mouse cell line that has transcriptional characteristics of both smooth and skeletal muscle lineages. Actively growing BC3H1 cells exhibit Cnn1 mRNA expression, which is extinguished when these cells are induced to differentiate upon serum withdrawal. Replating differentiated BC3H1 cells restores steady-state Cnn1 mRNA levels. The down-regulation of Cnn1 mRNA during BC3H1 differentiation coincides with the induction of myogenin, a skeletal muscle transcription factor that is not present in SMC lineages. Results from cycloheximide and actinomycin D studies suggest the existence of a labile repressor protein(s) that destabilizes the pool of Cnn1 mRNA and/or silences transcription of the Cnn1 locus. Mapping of the mouse Cnn1 locus to Chr 9, which is homologous to human Cnn1 on 19p13.2 and rat Cnn1 on 8q, suggests no gross rearrangement of this locus in the BC3H1 cell line. These results are the first to show reversible expression of Cnn1 and demonstrate the utility of the BC3H1 muscle cell line as a model system for the further characterization of Cnn1 gene regulation.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas Musculares/genética , Músculo Liso , Animais , Linhagem Celular , Mapeamento Cromossômico , Expressão Gênica , Cariotipagem , Camundongos , Proteínas dos Microfilamentos , Músculo Esquelético/fisiologia , RNA Mensageiro , Calponinas
6.
Biochem Biophys Res Commun ; 281(2): 475-82, 2001 Feb 23.
Artigo em Inglês | MEDLINE | ID: mdl-11181072

RESUMO

A modified suppression subtractive hybridization assay was performed to uncover genes induced by all-trans retinoic acid in cultured smooth muscle cells (SMC). Northern blotting studies confirmed the induction of 14 genes, many of which have heretofore been unrecognized as retinoid-inducible. Temporal expression and cycloheximide studies allowed us to categorize these genes as either immediate-early (LOX-1, endolyn, Stoned B/TFIIA alpha/beta-like factor, Src Suppressed C Kinase Substrate, and tissue transglutaminase) or delayed (cathepsin-L, ceruloplasmin, epithelin, importin alpha, alpha(8)-integrin, lactate dehydrogenase B, retinol dehydrogenase, spermidine/spermine N(1)-acetyltransferase, and VCAM-1) retinoid-response genes. A survey of rat tissues showed two of the genes (tissue transglutaminase and alpha(8)-integrin) to be highly restricted to vascular tissue. In situ hybridization verified expression of both tissue transglutaminase and alpha(8)-integrin to SMC in balloon-injured rat carotid artery. These findings unveil a new retinoid-response gene set that should be exploited to define molecular pathways involved in the antagonistic effects of retinoids on SMC growth and neointimal formation.


Assuntos
Regulação da Expressão Gênica/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Retinoides/farmacologia , Animais , Northern Blotting , Artérias Carótidas/efeitos dos fármacos , Artérias Carótidas/metabolismo , Artérias Carótidas/patologia , Células Cultivadas , Cicloeximida/farmacologia , Hibridização In Situ , Masculino , Músculo Liso Vascular/citologia , Músculo Liso Vascular/metabolismo , Hibridização de Ácido Nucleico/métodos , Inibidores da Síntese de Proteínas/farmacologia , RNA/efeitos dos fármacos , RNA/genética , RNA/metabolismo , RNA Mensageiro/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Fatores de Tempo , Distribuição Tecidual , Tretinoína/farmacologia
7.
Microcirculation ; 8(6): 403-13, 2001 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-11781813

RESUMO

OBJECTIVE: Our purpose was to develop a method for adenovirus delivery to the hamster cheek pouch to experimentally target gene transfer in tissue used for microvascular studies. METHODS: Separate constructs were tested with transgenes for lacZ or green fluorescent protein (GFP) driven by three promoters: RSV, CMV, and SM22. With university approval, adenovirus was delivered in anesthetized (pentobarbital, 70 mg/kg) hamsters (n = 28) by using either a vascular systemic injection or tissue infiltration (interstitial space behind the pouch). During 3 days, animals receiving infiltration gained the expected weight, whereas those receiving vascular injection lost weight; no other behavior changes were noted. RESULTS: On day 3 postadenoviral delivery (infiltration), expression of lacZ (histology, beta-galactosidase) or GFP (fluorescence microscopy) was confirmed across the tissue (CMV and RSV promoters) and exclusively in vascular smooth muscle cells (specific SM22 promoter), without evidence of tissue inflammation. In vitro microvascular experiments verified normal responses in the cheek pouch of day 3 postadenoviral delivery animals. We tested local dilation to methacholine, adenosine, remote dilation to methacholine, adenosine, nitroprusside, and LM609 (alpha(v)beta3 integrin agonist), flow-dependent dilation, and flow recruitment. CONCLUSIONS: Thus, this method enables targeted, cell-specific gene transfer to one tissue important for microvascular studies, without significant systemic exposure and without adverse inflammation.


Assuntos
Adenoviridae/genética , Técnicas de Transferência de Genes , Vetores Genéticos , Músculo Liso Vascular/metabolismo , Animais , Bochecha , Cricetinae , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/farmacocinética , Vetores Genéticos/toxicidade , Proteínas de Fluorescência Verde , Proteínas Luminescentes/genética , Proteínas Luminescentes/metabolismo , Proteínas Luminescentes/farmacocinética , Masculino , Mesocricetus , Microcirculação , Regiões Promotoras Genéticas , beta-Galactosidase/genética , beta-Galactosidase/metabolismo , beta-Galactosidase/farmacocinética
8.
Circ Res ; 87(10): 881-7, 2000 Nov 10.
Artigo em Inglês | MEDLINE | ID: mdl-11073883

RESUMO

Retinoids exert antiproliferative and prodifferentiating effects in vascular smooth muscle cells (SMCs) and reduce neointimal mass in balloon-injured blood vessels. The mechanisms through which retinoids carry out these effects are unknown but likely involve retinoid receptor-mediated changes in gene expression. Here we report the cloning, chromosomal mapping, and biological activity of the retinoid-response gene rat tissue transglutaminase (tTG). Northern blotting studies showed that tTG is rapidly and dose-dependently induced in a protein synthesis-independent manner after stimulation with the natural retinoid all-trans retinoic acid (atRA). The induction of tTG was selective for atRA and its stereoisomers 9-cis and 13-cis RA, because little or no elevation in mRNA expression was observed with a panel of growth factors. Western blotting and immunofluorescence confocal microscopy showed an accumulation of cytosolic tTG protein after atRA stimulation. Radiolabeled cross-linking studies revealed a corresponding elevation in in vitro tTG activity. The increase in tTG activity was reduced in the presence of 2 distinct inhibitors of tTG (monodansylcadaverine and cystamine). atRA-induced tTG mRNA and protein expression were followed by a significant elevation in SMC apoptosis. Such retinoid-induced programmed cell death could be partially inhibited with each tTG inhibitor and was completely blocked when both inhibitors were used simultaneously. These results establish a role for atRA in the sequential stimulation of tTG and apoptosis in cultured SMCs. atRA-mediated apoptosis in SMCs seems to require the participation of active tTG, suggesting a potential mechanistic link between this retinoid-inducible gene and programmed cell death.


Assuntos
Apoptose , Cadaverina/análogos & derivados , Proteínas de Ligação ao GTP/genética , Proteínas de Ligação ao GTP/metabolismo , Músculo Liso Vascular/metabolismo , Transglutaminases/genética , Transglutaminases/metabolismo , Tretinoína/metabolismo , Animais , Northern Blotting , Western Blotting , Cadaverina/farmacologia , Células Cultivadas , Mapeamento Cromossômico , Clonagem Molecular , Cistamina/farmacologia , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Proteínas de Ligação ao GTP/antagonistas & inibidores , Substâncias de Crescimento/metabolismo , Substâncias de Crescimento/farmacologia , Masculino , Dados de Sequência Molecular , Músculo Liso Vascular/citologia , Proteína 2 Glutamina gama-Glutamiltransferase , RNA Mensageiro/metabolismo , Ratos , Ratos Sprague-Dawley , Análise de Sequência de DNA , Homologia de Sequência de Aminoácidos , Transcrição Gênica/efeitos dos fármacos , Transglutaminases/antagonistas & inibidores , Tretinoína/farmacologia
10.
J Biol Chem ; 275(39): 30387-93, 2000 Sep 29.
Artigo em Inglês | MEDLINE | ID: mdl-10866994

RESUMO

Prolonged serum deprivation induces a structurally and functionally contractile phenotype in about 1/6 of cultured airway myocytes, which exhibit morphological elongation and accumulate abundant contractile apparatus-associated proteins. We tested the hypothesis that transcriptional activation of genes encoding these proteins accounts for their accumulation during this phenotypic transition by measuring the transcriptional activities of the murine SM22 and human smooth muscle myosin heavy chain promoters during transient transfection in subconfluent, serum fed or 7 day serum-deprived cultured canine tracheal smooth muscle cells. Contrary to our expectation, SM22 and smooth muscle myosin heavy chain promoter activities (but not viral murine sarcoma virus-long terminal repeat promoter activity) were decreased in long term serum-deprived myocytes by at least 8-fold. Because serum response factor (SRF) is a required transcriptional activator of these and other smooth muscle-specific promoters, we evaluated the expression and function of SRF in subconfluent and long term serum-deprived cells. Whole cell SRF mRNA and protein were maintained at high levels in serum-deprived myocytes, but SRF transcription-promoting activity, nuclear SRF binding to consensus CArG sequences, and nuclear SRF protein were reduced. Furthermore, immunocytochemistry revealed extranuclear redistribution of SRF in serum-deprived myocytes; nuclear localization of SRF was restored after serum refeeding. These results uncover a novel mechanism for physiological control of smooth muscle-specific gene expression through extranuclear redistribution of SRF and consequent down-regulation of its transcription-promoting activity.


Assuntos
Núcleo Celular/metabolismo , Proteínas de Ligação a DNA/metabolismo , Proteínas dos Microfilamentos/genética , Proteínas Musculares/genética , Músculo Liso/fisiologia , Cadeias Pesadas de Miosina/genética , Proteínas Nucleares/metabolismo , Animais , Transporte Biológico , Compartimento Celular , Meios de Cultura Livres de Soro , Citoplasma/metabolismo , Proteínas de Ligação a DNA/isolamento & purificação , Cães , Regulação para Baixo , Regulação da Expressão Gênica , Músculo Liso/citologia , Regiões Promotoras Genéticas , Fator de Resposta Sérica , Traqueia/citologia , Fator de Transcrição AP-2 , Fatores de Transcrição/isolamento & purificação
11.
J Biol Chem ; 275(13): 9814-22, 2000 Mar 31.
Artigo em Inglês | MEDLINE | ID: mdl-10734136

RESUMO

Smooth muscle calponin is a multifunctional, thin filament-associated protein whose expression is restricted to smooth muscle cell lineages in developing and postnatal tissues. Although the physiology of smooth muscle calponin has been studied extensively, the cis-elements governing its restricted pattern of expression have yet to be identified. Here we report on smooth muscle-specific enhancer activity within the first intron of smooth muscle calponin. Sequence analysis revealed a proximal consensus intronic CArG box and two distal intronic CArG-like elements, each of which bound recombinant serum response factor (SRF) as well as immunoreactive SRF from smooth muscle nuclear extracts. Site-directed mutagenesis studies suggested that the consensus CArG box mediates much of the intronic enhancer activity; mutating all three CArG elements abolished the ability of SRF to confer enhancer activity on the smooth muscle calponin promoter. Cotransfecting a dominant-negative SRF construct attenuated smooth muscle-specific enhancer activity, and transducing smooth muscle cells with adenovirus harboring the dominant-negative SRF construct selectively reduced steady-state expression of endogenous smooth muscle calponin. These results demonstrate an important role for intronic CArG boxes and the SRF protein in the transcriptional control of smooth muscle calponin in vitro.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Proteínas de Ligação a DNA/fisiologia , Regulação da Expressão Gênica/fisiologia , Músculo Liso/metabolismo , Proteínas Nucleares/fisiologia , Animais , Sequência de Bases , Células Cultivadas , DNA , Proteínas de Ligação a DNA/metabolismo , Elementos Facilitadores Genéticos , Íntrons , Masculino , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Músculo Liso/citologia , Proteínas Nucleares/metabolismo , Ligação Proteica , Ratos , Ratos Sprague-Dawley , Fator de Resposta Sérica , Calponinas
12.
Mamm Genome ; 11(2): 115-9, 2000 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-10656925

RESUMO

Smooth muscle cells (SMC) express a battery of lineage-restricted genes whose encoded proteins impart the unique contractile phenotype that characterizes this muscle type. While the encoded function of many SMC-restricted genes has been extensively analyzed, less is known about their position within the genome and the regulatory factors governing their transcription. In this report, we define the gene structure, 5' promoter analysis, and chromosomal mapping of the rat smooth muscle calponin (CnnI) gene. The rat CnnI gene is comprised of seven exons spanning approximately 8 kb of genomic sequence. The intron-exon boundaries of the rat CnnI gene match precisely those in human and mouse. Primer extension and RNase protection assays indicate two major transcription start positions (tsp). Comparative sequence analysis of the 5' promoter region reveals several conserved cis regulatory elements, including a TA-rich element within 30 nt of the tsp that could be a recognition site for TATA-binding protein and two CCAAT boxes. Transient and stable transfection studies support the hypothesis that distal regulatory elements confer SMC-restricted expression of CnnI. Finally, using an F2 intercross, we have mapped the rat CnnI gene to the telomeric end of Chromosome (Chr) 8. These studies provide additional information relating to the control of CnnI gene expression and provide a platform to begin assessing the potential linkage of CnnI to spontaneous and experimental disease phenotypes in rats.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Mapeamento Cromossômico , Proteínas Musculares/genética , Músculo Liso Vascular/química , Animais , Sequência de Bases , Proteínas de Ligação ao Cálcio/química , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Proteínas Musculares/química , Regiões Promotoras Genéticas , Ratos , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Calponinas
13.
Dev Biol ; 217(2): 301-9, 2000 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-10625555

RESUMO

The zinc finger transcription factors GATA4, -5, and -6 and the homeodomain protein Nkx2.5 are expressed in the developing heart and have been shown to activate a variety of cardiac-specific genes. To begin to define the regulatory relationships between these cardiac transcription factors and to understand the mechanisms that control their expression during cardiogenesis, we analyzed the mouse GATA6 gene for regulatory elements sufficient to direct cardiac expression during embryogenesis. Using beta-galactosidase fusion constructs in transgenic mice, a 4.3-kb 5' regulatory region that directed transcription specifically in the cardiac lineage, beginning at the cardiac crescent stage, was identified. Thereafter, transgene expression became compartmentalized to the outflow tract, a portion of the right ventricle, and a limited region of the common atrial chamber of the embryonic heart. Further dissection of this regulatory region identified a 1.8-kb cardiac-specific enhancer that recapitulated the expression pattern of the larger region when fused to a heterologous promoter and a smaller 500-bp subregion that retained cardiac expression, but was quantitatively weaker. The GATA6 cardiac enhancer contained a binding site for Nkx2.5 that was essential for cardiac-specific expression in transgenic mice. These studies demonstrate that GATA6 is a direct target gene for Nkx2.5 in the developing heart and reveal a mutually reinforcing regulatory network of Nkx2.5 and GATA transcription factors during cardiogenesis.


Assuntos
Proteínas de Ligação a DNA/genética , Elementos Facilitadores Genéticos , Regulação da Expressão Gênica no Desenvolvimento , Coração/embriologia , Proteínas de Homeodomínio/metabolismo , Fatores de Transcrição/genética , Proteínas de Xenopus , Animais , Sequência de Bases , Sítios de Ligação , Linhagem da Célula , Fator de Transcrição GATA6 , Proteína Homeobox Nkx-2.5 , Camundongos , Camundongos Transgênicos , Dados de Sequência Molecular , Regiões Promotoras Genéticas , Ligação Proteica , Ativação Transcricional
15.
Circ Res ; 84(10): 1166-76, 1999 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-10347091

RESUMO

A hallmark of vascular lesions is the phenotypic modulation of vascular smooth muscle cells (VSMCs) from a quiescent, contractile state to a more primitive, proliferative phenotype with a more fetal pattern of gene expression. Using subtraction hybridization to identify genes that may regulate this transition, we cloned a novel gene named EVEC, an acronym for its expression in the embryonic vasculature and the presence of Ca2+ binding epidermal growth factor-like repeats contained in the predicted protein structure. Although these repeats are characteristic of the extracellular matrix proteins, fibrillin, fibulin, and the latent transforming growth factor-beta binding proteins, EVEC most closely resembles the H411 and T16/S1-5 gene products, the latter of which are believed to regulate DNA synthesis in quiescent fibroblasts. Using in situ hybridization, we demonstrated that EVEC is expressed predominantly in the VSMCs of developing arteries in E11.5 through E16.5 mouse embryos. Lower levels of expression are also observed in endothelial cells, perichondrium, intestine, and mesenchyme of the face and kidney. EVEC mRNA expression is dramatically downregulated in adult arteries, except in the uterus, where cyclic angiogenesis continues; however, EVEC expression is reactivated in 2 independent rodent models of vascular injury. EVEC mRNA is observed in cellular elements of atherosclerotic plaques of LDL receptor-deficient, human apolipoprotein B transgenic mice and in VSMCs of the media and neointima of balloon-injured rat carotid arteries. These data suggest that EVEC may play an important role in the regulation of vascular growth and maturation during development and in lesions of injured vessels.


Assuntos
Fator de Crescimento Epidérmico/genética , Proteínas da Matriz Extracelular , Regulação da Expressão Gênica no Desenvolvimento , Músculo Liso Vascular/química , Proteínas Recombinantes , Fatores Etários , Animais , Arteriosclerose/genética , Arteriosclerose/metabolismo , Northern Blotting , Células COS , Células Cultivadas , Clonagem Molecular , Grânulos Citoplasmáticos/metabolismo , Elastina/análise , Fator de Crescimento Epidérmico/metabolismo , Feto/química , Feto/fisiologia , Hibridização In Situ , Camundongos , Microssomos/química , Microssomos/metabolismo , Dados de Sequência Molecular , Músculo Liso Vascular/citologia , Músculo Liso Vascular/fisiologia , Fenótipo , RNA Mensageiro/análise , Ratos , Sequências Repetitivas de Ácido Nucleico , Homologia de Sequência de Aminoácidos , Túnica Íntima/química , Túnica Íntima/citologia , Túnica Íntima/fisiologia , Regulação para Cima/fisiologia
16.
Methods Mol Med ; 30: 25-35, 1999.
Artigo em Inglês | MEDLINE | ID: mdl-21341013

RESUMO

Recent molecular genetic studies in cardiac and skeletal muscle have revealed mutations in a battery of sarcomeric muscle-restricted genes that appear to be associated with various myopathies (1,2). In sharp contrast, no mutations in smooth muscle cell (SMC)-restricted genes have been linked to a SMC disease phenotype, although a review of the literature indicates that many SMC diseases with a presumed genetic basis are present in human populations (3-13). An important first step in linking a disease phenotype to a mutation within a specific gene is the accurate physical mapping of the candidate gene to a specific chromosomal region within the context of other genetic markers, such as highly polymorphic microsatellite markers now routinely used for recombination-based linkage analysis of families segregating a particular disease phenotype. Several methods exist for the physical mapping of genes, including fluorescent in situ hybridization (FISH) (14) and interspecific mouse back-crossing (15). FISH analysis is relatively fast, but often requires large genomic clones and does not afford the high-resolution mapping required to link a gene locus to a disease phenotype. Interspecific mouse back-crossing can be quite powerful with respect to resolution, but studies are necessarily limited to the mouse genome. Thus, a broadly applicable, fast and simple method of gene mapping would be desirable to aid investigators in localizing potential candidate disease genes, especially those pertaining to SMC-associated diseases.

17.
Circulation ; 98(12): 1219-27, 1998 Sep 22.
Artigo em Inglês | MEDLINE | ID: mdl-9743514

RESUMO

BACKGROUND: The multifactorial and unpredictable nature of human restenosis will probably necessitate interventional strategies that target multiple processes involved in acute vascular narrowing. Retinoids (eg, all-trans-retinoic acid, atRA) represent a growing class of pleiotropic biological response modifiers with demonstrable efficacy in managing several pathological conditions. In this report, we have initiated studies to examine the hypothesis that atRA limits neointimal formation after experimental vascular injury. METHODS AND RESULTS: Rats were predosed with atRA (30 mg . kg-1 . d-1 PO) or corn oil 4 days before balloon withdrawal injury (BWI) of the left common carotid artery and continued on this drug regimen for an additional 14 days. High-performance liquid chromatographic analysis documented therapeutic levels of atRA in serum and vascular tissue. atRA depressed peak DNA synthesis in the tunica media of BWI vessels (P<0.05). Histomorphometry revealed atRA-mediated reductions in neointimal area, neointimal cell number, and intimal/medial area ratio as well as significant increases in vessel wall perimeter (P<0. 05). Such changes in vascular architecture contributed to a 35% to 37% increase in the luminal area of BWI vessels exposed to atRA (P<0. 005 compared with controls). CONCLUSIONS: atRA reduces neointimal mass and elicits favorable geometric remodeling of the injured rat carotid artery.


Assuntos
Artérias Carótidas/efeitos dos fármacos , Músculo Liso Vascular/efeitos dos fármacos , Tretinoína/farmacologia , Actinas/análise , Actinas/imunologia , Animais , Artérias Carótidas/patologia , DNA/biossíntese , Masculino , Músculo Liso Vascular/patologia , Ratos , Ratos Sprague-Dawley , Tretinoína/farmacocinética
18.
In Vitro Cell Dev Biol Anim ; 34(3): 217-26, 1998 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-9557939

RESUMO

Transcriptional regulation of smooth muscle cell (SMC) differentiation is a rapidly growing area of interest that has relevance for understanding intimal disease. Despite the wealth of data accumulating in vitro, however, no study has compared the cell-specific marker profile, transfectability, promoter activity, and growth characteristics among several SMC culture systems. Accordingly, we performed a comprehensive analysis of the marker profile, growth properties, transfectability, and SMC promoter activity in four rat SMC lines (A7r5, adult and pup aortic, and PAC1). Despite alterations in chromosomal number and structure, A7r5, adult aortic, and PAC1 cells express all SMC markers studied including SM alpha-actin, SM calponin, SM22, tropoelastin, and to a lesser extent, SM myosin heavy chain (SMMHC). In contrast, pup aortic cells express very low or undetectable levels of all the above markers except tropoelastin. Adult aortic, pup, and PAC1 cells display similar growth curves and levels of proto-oncogene transcripts, whereas those in the A7r5 line are comparatively less. All cell lines studied except pup cells show expression of SMC differentiation genes during active growth, indicating that growth and differentiation are not mutually exclusive in cultured smooth muscle. Transfection studies reveal dramatic differences in DNA uptake and SMC-restricted promoter activity between cell lines. Collectively, these results provide detailed information relating to SMC molecular biology in culture that should facilitate the selection of a cell line for studying the transcriptional regulatory mechanisms underlying SMC differentiation.


Assuntos
Músculo Liso Vascular/metabolismo , Actinas/análise , Animais , Biomarcadores , Proteínas de Ligação ao Cálcio/análise , Divisão Celular , Linhagem Celular , Proteínas dos Microfilamentos , Músculo Liso Vascular/citologia , Cadeias Pesadas de Miosina/análise , Regiões Promotoras Genéticas , Ratos , Ratos Endogâmicos WKY , Ratos Sprague-Dawley , Soroalbumina Bovina/farmacologia , Transfecção , Calponinas
20.
Gene ; 197(1-2): 215-24, 1997 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-9332369

RESUMO

Smooth muscle cells (SMC) express a battery of cell-restricted differentiation genes, many of which are down-regulated during the course of vascular disease. Here, we present the mRNA expression, genomic structure and chromosomal mapping of the gene encoding human smooth muscle cell calponin (SMCC). Human SMCC transcripts are restricted to tissues and cells of SMC origin and, in the latter case, appear to be uniquely controlled in two distinct human SMC lines of uterine and aortic origin. Restriction mapping. Southern blot and PCR analysis of a 70-kb human bacterial artificial chromosome (BAC) revealed a genomic structure (seven exons spanning > 11 kb) very similar to that reported for the mouse SMCC gene. Using a variety of human-rodent somatic cell hybrid and radiation hybrid mapping panels, the human SMCC gene was mapped to a genomic interval of less than 1.32 Mb in 19p13.2. These results provide new information concerning the regulation of SMCC gene expression and demonstrate the utility of two human SMC lines for the further characterization of this gene's expression control. The identification of a BAC harboring the entire human SMCC locus represents an important reagent for future analysis of SMCC regulatory sequences. Finally, the localization of SMCC to a defined genomic interval will facilitate an analysis of its potential as a candidate gene for disease phenotypes mapping to 19p13.2.


Assuntos
Proteínas de Ligação ao Cálcio/genética , Mapeamento Cromossômico , Cromossomos Humanos Par 19/genética , Regulação da Expressão Gênica/fisiologia , Proteínas Musculares/genética , Sequência de Aminoácidos , Sequência de Bases , Linhagem Celular , Clonagem Molecular/métodos , Éxons/genética , Genes/genética , Humanos , Proteínas dos Microfilamentos , Dados de Sequência Molecular , Músculo Liso/citologia , Especificidade de Órgãos , RNA Mensageiro/análise , Homologia de Sequência do Ácido Nucleico , Calponinas
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...